ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Structure-Based Discovery of A2A Adenosine Receptor Ligands

View Author Information
Department of Pharmaceutical Chemistry, University of California, 1700 4th Street, Box 2550, San Francisco, California 94158
Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
*To whom correspondence should be addressed. For B.K.S.: phone, 415-514-4126; fax, 415-514-4260; e-mail, [email protected]. For K.A.J.: phone, 301-496-9024; fax, 301-480-8422; e-mail, [email protected]
Cite this: J. Med. Chem. 2010, 53, 9, 3748–3755
Publication Date (Web):April 20, 2010
https://doi.org/10.1021/jm100240h

Copyright © 2010 American Chemical Society. This publication is licensed under these Terms of Use.

  • Open Access

Article Views

7321

Altmetric

-

Citations

LEARN ABOUT THESE METRICS
PDF (2 MB)
Supporting Info (1)»

Abstract

The recent determination of X-ray structures of pharmacologically relevant GPCRs has made these targets accessible to structure-based ligand discovery. Here we explore whether novel chemotypes may be discovered for the A2A adenosine receptor, based on complementarity to its recently determined structure. The A2A adenosine receptor signals in the periphery and the CNS, with agonists explored as anti-inflammatory drugs and antagonists explored for neurodegenerative diseases. We used molecular docking to screen a 1.4 million compound database against the X-ray structure computationally and tested 20 high-ranking, previously unknown molecules experimentally. Of these 35% showed substantial activity with affinities between 200 nM and 9 μM. For the most potent of these new inhibitors, over 50-fold specificity was observed for the A2A versus the related A1 and A3 subtypes. These high hit rates and affinities at least partly reflect the bias of commercial libraries toward GPCR-like chemotypes, an issue that we attempt to investigate quantitatively. Despite this bias, many of the most potent new ligands were novel, dissimilar from known ligands, providing new lead structures for modulation of this medically important target.

Introduction

ARTICLE SECTIONS
Jump To

G-protein-coupled receptors (GPCRsa)
a

Abbreviations: GPCR, G-protein-coupled receptor; AR, adenosine receptor; CNS, central nervous system; SEA, similarity ensemble approach; PDB, Protein Data Bank; WOMBAT, World of Molecular Bioactivity; CHO, Chinese hamster ovary; DMEM, Dulbecco’s modified Eagle medium; PKA, protein kinase A; DLS, dynamic light scattering.

are a large family of transmembrane proteins that signal intracellularly after binding an extracellular ligand. These receptors share a similar topology, with seven transmembrane helices, but recognize a wide range of different signaling molecules. GPCRs have been intensely studied as pharmaceutical targets, and over 40% of marketed drugs act through them. (1) Until recently, a missing link to deeper understanding of GPCRs has been a lack of atomic resolution structural information. With the recent advent of several X-ray crystal structures of pharmacologically relevant GPCRs (2-5) it has for the first time become possible to leverage high-resolution structures for ligand discovery against these targets. (6)
Among the new GPCR structures is that of the A2A adenosine receptor (AR). (5) There are four subtypes of the AR (A1, A2A, A2B, and A3), and they are activated by extracellular adenosine in response to organ stress or tissue damage. The A2A AR signals in both the periphery and the CNS, with agonists explored as anti-inflammatory drugs and antagonists explored for neurodegenerative diseases, e.g., Parkinson’s disease. (7-11) Although access to high resolution structural data is a crucial step toward atomic-level understanding of GPCRs, the lack of structures has certainly not been an obstacle for successful ligand discovery. For several decades, classical ligand-based medicinal chemistry approaches have been used to identify thousands of AR ligands. Almost all known AR agonists are derivatives of the cognate ligand (13, Chart 1), whereas antagonists are more diverse. Two large classes of AR antagonists are xanthines, with members such as caffeine (4) and theophylline (5), and adenine derivates such as 6 (ZM241385 (12)), which is bound to the A2A AR binding site in the crystallographic structure (Chart 1, Figure 1A). Despite considerable medicinal chemistry efforts and the wide range of possible therapeutic applications for AR ligands, there are only a few approved drugs targeting this receptor. (8, 11) Consequently, there remains an ongoing need for new subtype selective agonists and antagonists of this target.

Chart 1

Chart 1. Structures of Known Agonists (13) and Antagonists (46) of the A2A Adenosine Receptor

Figure 1

Figure 1. Binding mode of the cocrystallized ligand 6 (A) and the predicted binding modes of the seven ligands discovered in the docking screen (B−H). The A2A AR binding site is shown in white ribbons with the side chains of Glu169 and Asn253 in sticks. In (A) the cocrystallized ligand 6 is shown using orange carbon atoms. In (B−H), the crystallographic ligand is shown using blue lines and the docking poses for the ligands are depicted with orange carbon atoms. Black dotted lines indicate hydrogen bonds. The compounds are (B) 7, (C) 8, (D) 9, (E) 10, (F) 11, (G) 12, and (H) 13.

Here, we wished to investigate whether we could find new A2A AR ligand chemotypes by using structure-based molecular docking to screen a large and putatively unbiased library of small molecules, looking for those that complement the receptor structure. Docking evaluates the complementarity of small molecules to a receptor binding site of known structure (13-18) and can in principle discover new chemotypes, dissimilar to previous ligands, that nevertheless fit the binding site well. Such chemotypes might provide new routes for modulation of this key target. Methodologically, we wanted to explore what the hit rate of a structure-based (docking) screen against the A2A AR might be. In docking screens against the β2 adrenergic GPCR, a hit rate of 24% had been observed. (19-23) A docking “hit” is a molecule that binds to the target at a relevant concentration, and a docking “hit rate” is the number of compounds that bind divided by the number of compounds experimentally tested. For the β2 adrenergic receptor, where the affinity of the best docking hit was 9 nM, both were unusually high. We wished to understand whether this would be true for this second GPCR and why this might be so. To investigate this, we docked a library of 1.4 million small molecules to the crystal structure of the A2A AR. From the top-scoring molecules, 20 were selected on the basis of their fit to the binding site and chemical diversity. Here, we present the experimental evaluation of these molecules and assess why GPCRs appear to be particularly suitable targets for structure-based ligand discovery.

Methods

ARTICLE SECTIONS
Jump To

Preparation of the Molecular Docking Screen

All docking calculations were carried out with the program DOCK 3.5.54 (16, 17, 24) using a 2.6 Å crystallographic structure of the A2A adenosine receptor in complex with an antagonist (6) (12) (PDB accession code 3EML (5)). The receptor structure was prepared by removing all non-protein atoms and the intracellular T4-lysozyme insertion. The protonation states of ionizable residues were set to the most probable in aqueous solution at pH 7.
The flexible-ligand sampling algorithm in DOCK3.5.54 superimposes atoms of the docked molecule onto binding site matching spheres, which indicate putative ligand atom positions. (16, 17) In the case of the A2A AR, 45 matching spheres were used, and these were either based on the atoms of the crystallographic ligand or positioned manually. The spheres were also labeled for chemical matching based on the local receptor environment. (25) The degree of ligand sampling is determined by the bin size, bin size overlap, and distance tolerance. These three parameters were set to 0.4, 0.3, and 1.5 Å, respectively, for both the binding site matching spheres and the docked molecules. For ligand conformations passing an initial steric filter, a physics-based scoring function is used to evaluate the fit to the receptor binding site. For the best scoring conformation of each docked molecule, 100 steps of rigid-body minimization are carried out. The score for each conformation is calculated as the sum of the receptor−ligand electrostatic and van der Waals interaction energy, corrected for ligand desolvation. These three terms are evaluated from precalculated grids. The three-dimensional map of the electrostatic potential in the binding site was prepared using the program Delphi. (26) In this calculation, partial charges from the united atom AMBER force field (27) were used for all receptor atoms except the side chain amide of Asn253, for which the dipole moment was increased to favor hydrogen bonding to this residue (we have adopted this technique of increasing local dipoles on a few polar residues in the active site without changing their formal charges extensively in past studies). (28, 29) The program CHEMGRID was used to generate a van der Waals grid, which is based on a united atom version of the AMBER force field. (30) The desolvation penalty for a ligand conformation is estimated from a precalculated transfer free energy of the molecule between solvents of dielectrics 78 and 2. The desolvation energy is obtained by weighting the transfer free energy with a scaling factor that reflects the degree of burial of the ligand in the receptor binding site. (31, 32)
The ZINC leadlike set was prepared by filtering a large library of commercially available compounds using the criteria log P < 3.5, molecular weight of <350, and number of rotatable bonds of ≤7. (33) Each molecule has been prepared for docking by pregenerating up to 1000 conformations using the program OMEGA. (34) Partial atomic charges and transfer free energies have been calculated using AMSOL, (35, 36) and van der Waals parameters have been derived from an all-atom AMBER potential. (37)

Similarity and Library Bias Calculations

Similarity calculations were carried out with the program Pipeline Pilot (38) using the Tanimoto coefficient and ECFP4 fingerprints. For each of the docking-discovered ligands, the Tanimoto similarity to all annotated A1, A2A, A2B, and A3 AR ligands with Ki ≤ 10 μM in the World of Molecular Bioactivity (WOMBAT 2006.2) (39) and ChEMBL (a StARlite 2009 prerelease version) (40) databases was calculated. The number of molecules in the ZINC leadlike database that are similar to known ligands of the ARs, adrenergic receptors, adenylyl cyclases, and AmpC β-lactamase was predicted with the similarity ensemble approach (SEA) using ECFP4 fingerprints. (41) Ligands (Ki ≤ 10 μM) that are annotated to ARs, adrenergic receptors, and adenylyl cyclases were extracted from the WOMBAT database. Ligands for AmpC β-lactamase were extracted from refs 28, 29, and 42. For each ligand set, the number of leadlike molecules in ZINC that have a SEA P value better than 10−10 was calculated. The predicted compounds were then postfiltered for molecules that match the molecular weight and formal charge ranges of the known ligands.

A2A AR Receptor Binding and Functional Assay

Binding assays at three hAR subtypes were carried out using standard radioligands (43-45) and membrane preparations from Chinese hamster ovary (CHO) cells (A1 and A3) or human embryonic kidney (HEK293) cells (A2A) stably expressing a hAR subtype. (46, 47) A functional assay at the A2AAR consisted of stimulation of cAMP production (48, 49) in A2AAR-expressing HEK293 cells. [3H]R-N6-(2-phenylisopropyl)adenosine ([3H]R-PIA, 42.6 Ci/mmol) was obtained from Moravek Biochemicals (Brea, CA). [3H](2-[p-(2-Carboxyethyl)phenylethylamino]-5′-N-ethylcarboxamidoadenosine) ([3H]CGS21680, 40.5 Ci/mmol) and [125I]N6-(4-amino-3-iodobenzyl)adenosine-5′-N-methyluronamide ([125I]I-AB-MECA, 2200 Ci/mmol) were purchased from Perkin-Elmer Life and Analytical Science (Boston, MA). Test compounds were prepared as 5 mM stock solutions in DMSO and stored frozen at −20 °C.

Cell Culture and Membrane Preparation

CHO cells stably expressing the recombinant hA1 and hA3Rs, and HEK-293 cells stably expressing the hA2AAR were cultured in Dulbecco’s modified Eagle medium (DMEM) and F12 (1:1) supplemented with 10% fetal bovine serum, 100 units/mL penicillin, 100 μg/mL streptomycin, and 2 μmol/mL glutamine. In addition, 800 μg/mL Geneticin was added to the A2A media, while 500 μg/mL hygromycin was added to the A1 and A3 media. After being harvested, cells were homogenized and suspended in PBS. Cells were then centrifuged at 240g for 5 min, and the pellet was resuspended in 50 mM Tris-HCl buffer (pH 7.5) containing 10 mM MgCl2. The suspension was homogenized and was then ultracentrifuged at 14330g for 30 min at 4 °C. The resultant pellets were resuspended in Tris buffer and incubated with adenosine deaminase (3 units/mL) for 30 min at 37 °C. The suspension was homogenized with an electric homogenizer for 10 s, pipetted into 1 mL vials, and then stored at −80 °C until the binding experiments were conducted. The protein concentration was measured using the BCA protein assay kit from Thermo Scientific Pierce Protein Research Products (Rockford, IL). (50)

Binding Assays

The tested compounds were purchased from six different vendors (Enamine, ChemDiv, ChemBridge, Vitas-M, Pharmeks, and Asinex). The vendors had verified that each compound had ≥95% purity by liquid chromatography−mass spectrometry (LC−MS) or nuclear magnetic resonance (NMR) experiments. Into each tube in the binding assay was added 50 μL of increasing concentrations of the test ligand in Tris-HCl buffer (50 mM, pH 7.5) containing 10 mM MgCl2, 50 μL of the appropriate agonist radioligand, and finally 100 μL of membrane suspension. For the A1AR (22 μg of protein/tube) the radioligand used was [3H]R-PIA (final concentration of 3.5 nM). For the A2AAR (20 μg/tube) the radioligand used was [3H]CGS21680 (10 nM). For the A3AR (21 μg/tube) the radioligand used was [125I]I-AB-MECA (0.34 nM). Nonspecific binding was determined using a final concentration of 10 μM unlabeled 5′-N-ethylcarboxamidoadenosine (NECA, 2) diluted with the buffer. The mixtures were incubated at 25 °C for 60 min in a shaking water bath. Binding reactions were terminated by filtration through Brandel GF/B filters under a reduced pressure using a M-24 cell harvester (Brandel, Gaithersburg, MD). Filters were washed three times with 3 mL of 50 mM ice-cold Tris-HCl buffer (pH 7.5). Filters for A1 and A2AAR binding were placed in scintillation vials containing 5 mL of Hydrofluor scintillation buffer and counted using a Perkin Elmer liquid scintillation analyzer (Tri-Carb 2810TR). Filters for A3AR binding were counted using a Packard Cobra II γ-counter. The Ki values were determined using GraphPad Prism for all assays.

Cyclic AMP Accumulation Assay

Intracellular cyclic AMP (cAMP) levels were measured with a competitive protein binding method. (48, 49) CHO293 cells that expressed the recombinant human A2AAR were harvested by trypsinization. After centrifugation and resuspension in medium, cells were planted in 24-well plates in 1.0 mL of medium. After 24 h, the medium was removed and cells were washed three times with 1 mL of DMEM containing 50 mM HEPES, pH 7.4. Cells were then treated with the test compound in the presence of rolipram (10 μM) and adenosine deaminase (3 units/mL), and incubation was continued for an additional 1 h. The reaction was terminated by removing the supernatant, and cells were lysed upon the addition of 200 μL of 0.1 M ice-cold HCl. The cell lysate was resuspended and stored at −20 °C. For determination of cyclic AMP production, protein kinase A (PKA) was incubated with [3H]cyclic AMP (2 nM) in K2HPO4/EDTA buffer (K2HPO4, 150 mM; EDTA, 10 mM), 20 μL of the cell lysate, and 30 μL of 0.1 M HCl or 50 μL of cyclic AMP solution (0−16 pmol/200 μL for standard curve). Bound radioactivity was separated by rapid filtration through Whatman GF/C filters and washed once with cold buffer. Bound radioactivity was measured by liquid scintillation spectrometry.

Counterscreen for Colloidal Inhibition

To control for artifactual inhibition by colloidal aggregation, we looked for particle formation by Dynamic Light Scattering (DLS) and by inhibition of two counterscreen enzymes, cruzain and AmpC β-lactamase. Concentrated DMSO stocks of compounds were diluted with filtered 50 mM KPi, pH 7.0. Measurements were made using a DynaPro MS/X (Wyatt Technology) with a 55 mW laser at 826.6 nm. The laser power was 100%, and the detector angle was 90°. Cruzain assays were performed in 100 mM sodium acetate, pH 5.5, containing 5 mM DTT with and without 0.1% Triton X-100. Compounds were incubated with 0.8 nM cruzain for 5 min, and reactions were initiated by adding the fluorogenic substrate Z-Phe-Arg-aminomethylcoumarin (Z-FR-AMC). The final reaction volume was 200 μL with cruzain at 0.4 nM and ZF-R-AMC at 2.5 μM. Final DMSO concentrations were 0.5%. To measure enzyme inhibition, the increase in fluorescence (excitation wavelength of 355 nm, emission wavelength of 460 nm) was recorded for 5 min in a microtiter plate spectrofluorimeter (Molecular Devices, FlexStation). Assays were performed in duplicate in 96-well plates, with controls measuring enzyme activity in the presence of DMSO. Activity was measured for seven different concentrations for each compound. Inhibition of AmpC β-lactamase was measured for the two best compounds identified here, compounds 9 and 11, to complement the cruzain assay results. Assays were performed in 50 mM potassium phosphate, pH 7.0. Compounds were incubated with 1 nM β-lactamase for 5 min, and reactions were initiated by adding the substrate CENTA to a final concentration 92 μM. The final reaction volume was 1 mL. To measure enzyme inhibition, the increase in absorbance at 405 nm was recorded for 5 min in a UV−vis spectrophotometer (Agilent). Assays were performed in duplicate in 1 mL cuvettes, with controls measuring enzyme activity in the presence of DMSO. Activity was measured at 10 μM, 0.1% DMSO.

Results and Discussion

ARTICLE SECTIONS
Jump To

Molecular Docking Screen and Compound Selection

The program DOCK3.5.54 (16, 17) was used to screen 1.4 million commercially available “leadlike” molecules from the ZINC (33) database against the orthosteric site of the A2A AR. On average, each molecule was sampled in 5000 orientations and, for each that fit, an average of 16 500 conformations; the receptor was held rigid. Each molecule was scored for electrostatic and van der Waals complementarity corrected for ligand desolvation. Molecules typically overlapped with the geometry of the crystallized antagonist, making a mixture of polar and hydrophobic interactions, packing deeply into the site (Figure 1). The 500 top-ranking molecules (Table S1, Supporting Information), 0.035% of the docking prioritized library, were analyzed visually for features that are not taken into account in the docking calculation. This is a standard procedure for all our docking screens in which each of the molecules is inspected for novelty, physical properties, and binding energy contributions that are not included in the docking scoring function. For example, compound 9 (Table 1) was chosen both because it complemented the site well and because there were several molecules with the same scaffold in the top 500 list of compounds (e.g., the molecules ranked 7, 31, 59, 104, 115, 135, 137, 172, 184, 199, and 278 in Table S1, Supporting Information). From this evaluation, 20 compounds representing diverse molecules (within the context of the top 500-ranking molecules) were prioritized for experimental testing.
Table 1. Ligand Structures and Experimental Data for the Seven Hits from the Docking Screen against the A1, A2A, and A3 ARs
a

Rank in the docking screen.

b

Measured in three independent experiments.

c

The most similar compound annotated to ARs in WOMBAT (39) and ChEMBL. (40)

d

Tanimoto similarity coefficient to the closest annotated adenosine receptor ligand from ECFP4 fingerprints.

Radioligand Displacement Assays and Docking Hit Rate

The 20 compounds selected from the docking screen were tested for binding in a radioligand displacement assay. Seven of these molecules inhibited binding by ≥40% at 20 μM, corresponding to a “hit rate” of 35%. Subsequent dose−response curves were well-behaved, with Ki values varying from 200 nM to 8.8 μM (Table 1 and Figure 2). Four of the ligands, 9, 10, 11 and 13, were counterscreened for colloidal aggregation, a common mechanism of artifactual inhibition. (51) No colloidal particles were observed at 10 μM, by dynamic light scattering, for 9 and 13, nor did they inhibit cruzain at the same concentration. For compounds 10 and 11, particles were observed at 10 μM, but for 10 these appeared to be precipitant rather than colloids and this compound did not inhibit cruzain up to 10 μM. For compound 11 particles were observed at 10 μM, as was enzyme inhibition, but this inhibition was not reversible by detergent, inconsistent with colloidal aggregation. Furthermore, no inhibition of AmpC β-lactamase was observed up to 10 μM for 9 and 11. Taken together with the well-behaved dose−response curves (Figure 2), these results indicate that the molecules are well behaved, classical binding ligands.

Figure 2

Figure 2. Representative dose−response curves for displacement of binding of the radiolabeled A2A AR agonist 3 by compounds 9, 10, and 11.

All seven docking hits are specific for the A2A AR versus the related A1 and A3 subtypes (Table 1). Notably, our most potent ligand, compound 11, is also the most specific with over 50-fold higher affinity at the A2A AR. We therefore investigated five of its analogues not picked in the first round of docking that also fit well into the site (compounds 1418, Table 2). Four of the analogues were found to bind to the A2A AR with submicromolar affinities, and these molecules also had an improved A2A/A1 subtype selectivity. From these results, the prospects of identifying specific high-affinity A2A antagonists in this new class of compounds appear promising.
Table 2. Binding Affinities and Structures of Five Analogues to Compound 11 in Radioligand Binding Assays at A1, A2A, and A3 ARs
a

Measured in three independent experiments.

To put the results from this docking screen in perspective, our laboratory considers a high-throughput docking screen to be successful if a hit rate of 5% with ligand affinities in the micromolar range can be achieved. For example, we tested 56 compounds from a docking screen against AmpC β-lactamase and found one compound with a Ki value better than 100 μM, corresponding to a hit rate of 2%; (29) this inhibitor had a Ki value of 26 μM (Table 3). In the case of the A2A AR we observe 10-fold higher hit rate and the affinities of the hits are 10- to 100-fold better. Intriguingly, similar results were obtained in two docking screens against the other pharmaceutically relevant GPCR for which a crystallographic structure has been solved, the β2 adrenergic receptor. (19, 20) Kolb et al. identified six previously unknown ligands of the β2 adrenergic receptor, a 24% hit rate, with affinities as high as 9 nM. (19) It may be that GPCRs are particularly well-suited for structure-based docking screens, a point to which we will return.
Table 3. Target Library Bias and Docking Hit Rates
  representative DOCK screen
targetno. of ZINC molecules similar to known ligandsahit rateb (%)best potencyc (nM)
adenosine receptors424035200
adrenergic receptors4146249 (19)
adenylyl cyclases565450000 (55)
AmpC β-lactamase5452−526000 (29)
a

ZINC leadlike molecules with at least 10−10P values to annotated target ligands in WOMBAT using the similarity ensemble approach (SEA).

b

(Number of true ligands)/(number of predictions tested experimentally).

c

The affinity of the ligand with the best potency from the docking screen.

Predicted Binding Modes, Novelty, and Efficacy of the Discovered A2A AR Ligands

All seven of the new ligands are predicted to interact with the key recognition residue Asn253 in transmembrane helix 6 and many also hydrogen-bond with the carboxylate of Glu169 in extracellular loop 2, both in the orthosteric site of the receptor (Figure 1). The importance of interactions with Asn253 was identified early in our docking screens. We found that increasing the dipole moment of the Asn253 side chain amide, a technique we employ frequently, substantially increased the enrichment of known A2A AR ligands among a database of decoys in control calculations. Asn253 is conserved in all four AR receptor subtypes and has also been found to be a crucial interaction partner for both agonists and antagonists in mutagenesis studies. (52)
Whereas all the seven ligands are previously uncharacterized for the A2A AR, some of them bear known chemotypes. To quantify their novelty, or lack of it, we calculated the similarity of each molecule to 7500 known AR ligands from the WOMBAT and ChEMBL databases using pairwise Tanimoto coefficients (Tc, ECFP4 fingerprints) (Table 1). Ligand 12 resembles members of the xanthine class of antagonists, while compound 7 resembles certain quinazoline (53) ligands. Conversely, whereas compounds 10, 11, and 13 do conserve several moieties with known ligands, they also differ substantially from them, with Tc values of 0.3 to the closest annotated ligand. Nevertheless, they complement the site well both sterically and electrostatically (Figure 1). The potency of these molecules suggests that they may merit further study as new lead families for antagonists of the A2A AR.
To determine the efficacy of the compounds, their ability to inhibit intracellular cAMP production induced by agonist 3 was tested. No stimulation of cAMP production was detected for any of the molecules, while a clear displacement of agonist function was observed for the two most potent compounds, 9 and 11 (Figure 3). All seven of the new ligands are thus almost certainly A2A antagonists, as is the cocrystallized ligand. Intriguingly, this efficacy bias was also observed in the docking screens against the β2 adrenergic receptor, where only inverse agonists were found against the structure crystallized with the inverse agonist carazolol. In docking to rigid GPCR structures, the protein conformation may bias the screen toward molecules with the same efficacy as the cocrystallized ligand. This represents a challenge to our ability to exploit these structures for mechanisms of action, such as agonism, not represented in the experimental structure. To further investigate this, we determined the rank of two agonists, 1 and 2 (Chart 1), that were present in the set of commercially available molecules screened against the A2A AR binding site. Whereas multiple known antagonists would have been ranked among the top 500 molecules in the docking screen, these two agonists were ranked 951 057 and 919 993, respectively.

Figure 3

Figure 3. Functional assay based on measuring the production of cAMP for 3 (control), a potent A2A AR agonist, with or without 10 μM 9 or 11. The dose−response curve is shifted for both compounds, as expected in the case of competitive antagonistic inhibition. The % activation refers to production of cAMP normalized to the effect of 3 at 100 μM.

Is There Library Bias toward GPCR Chemotypes in Chemical Libraries?

Returning to one of the questions that motivated this study, the structure-based screen against the A2A AR returned a diverse set of ligands dissimilar to those previously characterized, as well as several similar to known ligands, and did so with a hit rate of 35%. Not only is this hit rate much higher than we have come to expect for enzyme targets screened with the same approach, but the new antagonists were also close to 100-fold more potent than we have come to expect for our docking “hits”. Furthermore, these results are strikingly similar to those observed in docking screens against the β2 adrenergic GPCR. (19, 20) To what may these unusually high hit rates and affinities be attributed?
Family A GPCRs like the A2A AR are the targets for a substantial fraction of marketed drugs, and this partly reflects the quality of their sites for specific recognition of small molecules. Largely buried from bulk solvent, these sites can almost completely enclose a “druglike” molecule and can do so with a mixture of nonpolar and polar interactions. Consequently, a large and sustained medicinal chemistry effort has focused on these targets, and by now even putatively unbiased libraries, like ZINC, have become populated with molecules bearing “GPCR-like” chemotypes. This also reflects a bias toward naturally occurring molecules in our screening libraries. (54) Indeed, Kolb et al. estimated that there were 3−12 times as many small molecules that were similar to GPCR ligands in the ZINC leadlike set compared to other common drug targets such as kinases, proteases, and ligand-gated ion channels. (19) To make this comparison more specific and relevant to the adenosine receptor versus other docking targets that we ourselves have worked, we investigated the library bias in ZINC for the adrenergic and adenosine receptors together with two other targets for which we have observed much lower hit rates and affinities (19, 29, 55) (Table 3). The number of molecules in the ZINC leadlike set that are similar to the ligands of these targets was estimated using SEA, (54) insisting on a P value of 10−10 or better; acceptable molecules had also to resemble the annotated ligands in their physical properties (see Methods). Over 4000 small molecules resemble ligands annotated to the ARs and adrenergic receptors in the WOMBAT (39) database, almost 10-fold more than found for the enzymes adenylyl cyclase and AmpC β-lactamase, against which, correspondingly, our docking hit rates and affinities have been 10- to 100-fold lower. Thus, it is the convolution of the high “ligand-ability” of the orthosteric sites and the many GPCR-like chemotypes in our libraries that makes the adrenergic and adenosine receptors so fruitful for structure-based techniques.

Note Added after Initial Review of This Paper

After this paper was submitted for review, a paper by Abagyan, Stevens, and colleagues appeared that also targeted the A2A AR for novel inhibitor discovery, also using a molecular docking screen. (56) As here, Abagyan et al. also observed a very substantial hit rate with high affinities (indeed, both the affinities and hit rates were slightly better than those we observe). Whereas the compounds discovered in the two screens were substantially different, the observation of the high hit rates and, by screening standards, high affinity “hits” is consistent with the high “ligand-ability” of the class A GPCRs and the fortuitous library bias toward them, and this is among the important conclusions of this study.

Supporting Information

ARTICLE SECTIONS
Jump To

Table S1 of structures of the 500 top-ranking molecules from the docking screen. This material is available free of charge via the Internet at http://pubs.acs.org.

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Authors
    • Brian K. Shoichet - Department of Pharmaceutical Chemistry, University of California, 1700 4th Street, Box 2550, San Francisco, California 94158 Email: [email protected] [email protected]
    • Kenneth A. Jacobson - Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892 Email: [email protected] [email protected]
  • Authors
    • Jens Carlsson - Department of Pharmaceutical Chemistry, University of California, 1700 4th Street, Box 2550, San Francisco, California 94158
    • Lena Yoo - Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
    • Zhan-Guo Gao - Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
    • John J. Irwin - Department of Pharmaceutical Chemistry, University of California, 1700 4th Street, Box 2550, San Francisco, California 94158

Acknowledgment

ARTICLE SECTIONS
Jump To

This work is supported by NIH Grant GM59957 (to B.K.S.), NIDDK Intramural Research Program (to K.A.J.), and a fellowship from the Knut and Alice Wallenberg Foundation (to J.C.). We thank A. Doak for aggregation assays and members of the Shoichet lab for docking “hit-list” evaluation. We thank Tudor Oprea for access to the WOMBAT database and John Overington for a prerelease version of the ChEMBL database.

References

ARTICLE SECTIONS
Jump To

This article references 56 other publications.

  1. 1
    Overington, J. P.; Al-Lazikani, B.; Hopkins, A. L. How many drug targets are there? Nat. Rev. Drug Discovery 2006, 5, 993 996
  2. 2
    Cherezov, V.; Rosenbaum, D. M.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Kuhn, P.; Weis, W. I.; Kobilka, B. K.; Stevens, R. C. High-resolution crystal structure of an engineered human beta(2)-adrenergic G protein-coupled receptor Science 2007, 318, 1258 1265
  3. 3
    Rosenbaum, D. M.; Cherezov, V.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Yao, X. J.; Weis, W. I.; Stevens, R. C.; Kobilka, B. K. GPCR engineering yields high-resolution structural insights into beta(2)-adrenergic receptor function Science 2007, 318, 1266 1273
  4. 4
    Warne, T.; Serrano-Vega, M. J.; Baker, J. G.; Moukhametzianov, R.; Edwards, P. C.; Henderson, R.; Leslie, A. G. W.; Tate, C. G.; Schertler, G. F. X. Structure of a beta(1)-adrenergic G-protein-coupled receptor Nature 2008, 454, 486 491
  5. 5
    Jaakola, V. P.; Griffith, M. T.; Hanson, M. A.; Cherezov, V.; Chien, E. Y. T.; Lane, J. R.; IJzerman, A. P.; Stevens, R. C. The 2.6 angstrom crystal structure of a human A(2A) adenosine receptor bound to an antagonist Science 2008, 322, 1211 1217
  6. 6
    Congreve, M.; Marshall, F. The impact of GPCR structures on pharmacology and structure-based drug design Br. J. Pharmacol. 2010, 159, 986 996
  7. 7
    Moro, S.; Gao, Z. G.; Jacobson, K. A.; Spalluto, G. Progress in the pursuit of therapeutic adenosine receptor antagonists Med. Res. Rev. 2006, 26, 131 159
  8. 8
    Jacobson, K. A.; Gao, Z. G. Adenosine receptors as therapeutic targets Nat. Rev. Drug Discovery 2006, 5, 247 264
  9. 9
    Sebastiao, A. M.; Ribeiro, J. A. Adenosine receptors and the central nervous system Handb. Exp. Pharmacol. 2009, 471 534
  10. 10
    Blackburn, M. R.; Vance, C. O.; Morschl, E.; Wilson, C. N. Adenosine receptors and inflammation Handb. Exp. Pharmacol. 2009, 215 269
  11. 11
    Cristalli, G.; Muller, C. E.; Volpini, R. Recent developments in adenosine A2A receptor ligands Handb. Exp. Pharmacol. 2009, 59 98
  12. 12
    Poucher, S. M.; Keddie, J. R.; Singh, P.; Stoggall, S. M.; Caulkett, P. W. R.; Jones, G.; Collis, M. G. The in-vitro pharmacology of Zm-241385, a potent, nonxanthine, a(2a) selective adenosine receptor antagonist Br. J. Pharmacol. 1995, 115, 1096 1102
  13. 13
    Degen, J.; Rarey, M. FlexNovo: structure-based searching in large fragment spaces ChemMedChem 2006, 1, 854 868
  14. 14
    Jones, G.; Willett, P.; Glen, R. C.; Leach, A. R.; Taylor, R. Development and validation of a genetic algorithm for flexible docking J. Mol. Biol. 1997, 267, 727 748
  15. 15
    Kairys, V.; Fernandes, M. X.; Gilson, M. K. Screening drug-like compounds by docking to homology models: a systematic study J. Chem. Inf. Model. 2006, 46, 365 379
  16. 16
    Lorber, D. M.; Shoichet, B. K. Flexible ligand docking using conformational ensembles Protein Sci. 1998, 7, 938 950
  17. 17
    Lorber, D. M.; Shoichet, B. K. Hierarchical docking of databases of multiple ligand conformations Curr. Top. Med. Chem. 2005, 5, 739 749
  18. 18
    Zavodszky, M. I.; Kuhn, L. A. Side-chain flexibility in protein−ligand binding: the minimal rotation hypothesis Protein Sci. 2005, 14, 1104 1114
  19. 19
    Kolb, P.; Rosenbaum, D. M.; Irwin, J. J.; Fung, J. J.; Kobilka, B. K.; Shoichet, B. K. Structure-based discovery of beta(2)-adrenergic receptor ligands Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 6843 6848
  20. 20
    Sabio, M.; Jones, K.; Topiol, S. Use of the X-ray structure of the beta(2)-adrenergic receptor for drug discovery. Part 2: Identification of active compounds Bioorg. Med. Chem. Lett. 2008, 18, 5391 5395
  21. 21
    de Graaf, C.; Rognan, D. Selective structure-based virtual screening for full and partial agonists of the beta 2 adrenergic receptor J. Med. Chem. 2008, 51, 4978 4985
  22. 22
    Katritch, V.; Reynolds, K. A.; Cherezov, V.; Hanson, M. A.; Roth, C. B.; Yeager, M.; Abagyan, R. Analysis of full and partial agonists binding to beta(2)-adrenergic receptor suggests a role of transmembrane helix V in agonist-specific conformational changes J. Mol. Recognit. 2009, 22, 307 318
  23. 23
    Reynolds, K. A.; Katritch, V.; Abagyan, R. Identifying conformational changes of the beta(2) adrenoceptor that enable accurate prediction of ligand/receptor interactions and screening for GPCR modulators J. Comput.-Aided Mol. Des. 2009, 23, 273 288
  24. 24
    Kuntz, I. D.; Blaney, J. M.; Oatley, S. J.; Langridge, R.; Ferrin, T. E. A geometric approach to macromolecule−ligand interactions J. Mol. Biol. 1982, 161, 269 288
  25. 25
    Shoichet, B. K.; Kuntz, I. D. Matching chemistry and shape in molecular docking Protein Eng. 1993, 6, 723 732
  26. 26
    Nicholls, A.; Honig, B. A rapid finite-difference algorithm, utilizing successive over-relaxation to solve the Poisson−Boltzmann equation J. Comput. Chem. 1991, 12, 435 445
  27. 27
    Weiner, S. J.; Kollman, P. A.; Case, D. A.; Singh, U. C.; Ghio, C.; Alagona, G.; Profeta, S.; Weiner, P. A new force-field for molecular mechanical simulation of nucleic-acids and proteins J. Am. Chem. Soc. 1984, 106, 765 784
  28. 28
    Babaoglu, K.; Simeonov, A.; Lrwin, J. J.; Nelson, M. E.; Feng, B.; Thomas, C. J.; Cancian, L.; Costi, M. P.; Maltby, D. A.; Jadhav, A.; Inglese, J.; Austin, C. P.; Shoichet, B. K. Comprehensive mechanistic analysis of hits from high-throughput and docking screens against beta-lactamase J. Med. Chem. 2008, 51, 2502 2511
  29. 29
    Powers, R. A.; Morandi, F.; Shoichet, B. K. Structure-based discovery of a novel, noncovalent inhibitor of AmpC beta-lactamase Structure 2002, 10, 1013 1023
  30. 30
    Meng, E. C.; Shoichet, B. K.; Kuntz, I. D. Automated docking with grid-based energy evaluation J. Comput. Chem. 1992, 13, 505 524
  31. 31
    Shoichet, B. K.; Leach, A. R.; Kuntz, I. D. Ligand solvation in molecular docking Proteins: Struct., Funct., Genet. 1999, 34, 4 16
  32. 32
    Wei, B. Q. Q.; Baase, W. A.; Weaver, L. H.; Matthews, B. W.; Shoichet, B. K. A model binding site for testing scoring functions in molecular docking J. Mol. Biol. 2002, 322, 339 355
  33. 33
    Irwin, J. J.; Shoichet, B. K. ZINC—a free database of commercially available compounds for virtual screening J. Chem. Inf. Model. 2005, 45, 177 182
  34. 34
    Bostrom, J.; Greenwood, J. R.; Gottfries, J. Assessing the performance of OMEGA with respect to retrieving bioactive conformations J. Mol. Graphics Modell. 2003, 21, 449 462
  35. 35
    Chambers, C. C.; Hawkins, G. D.; Cramer, C. J.; Truhlar, D. G. Model for aqueous solvation based on class IV atomic charges and first solvation shell effects J. Phys. Chem. 1996, 100, 16385 16398
  36. 36
    Li, J. B.; Zhu, T. H.; Cramer, C. J.; Truhlar, D. G. New class IV charge model for extracting accurate partial charges from wave functions J. Phys. Chem. A 1998, 102, 1820 1831
  37. 37
    Weiner, S. J.; Kollman, P. A.; Nguyen, D. T.; Case, D. A. An all atom force-field for simulations of proteins and nucleic-acids J. Comput. Chem. 1986, 7, 230 252
  38. 38
    http://accelrys.com/products/scitegic/.
  39. 39
    Olah, M.; Mracec, M.; Ostopovici, L.; Rad, R.; Bora, A.; Hadaruga, N.; Olah, I.; Banda, M.; Simon, Z.; Mracec, M.; Oprea, T. I. WOMBAT: World of Molecular Bioactivity. In Chemoinformatics in Drug Discovery; Oprea, T. I., Ed.; Wiley-VCH: Weinheim, Germany, 2005; pp 221239.
  40. 40
    http://www.ebi.ac.uk/chembl.
  41. 41
    Keiser, M. J.; Roth, B. L.; Armbruster, B. N.; Ernsberger, P.; Irwin, J. J.; Shoichet, B. K. Relating protein pharmacology by ligand chemistry Nat. Biotechnol. 2007, 25, 197 206
  42. 42
    Tondi, D.; Morandi, F.; Bonnet, R.; Costi, M. P.; Shoichet, B. K. Structure-based optimization of a non-beta-lactam lead results in inhibitors that do not up-regulate beta-lactamase expression in cell culture J. Am. Chem. Soc. 2005, 127, 4632 4639
  43. 43
    Jarvis, M. F.; Schulz, R.; Hutchison, A. J.; Do, U. H.; Sills, M. A.; Williams, M. [H-3] Cgs-21680, a selective A2 adenosine receptor agonist directly labels A2-receptors in rat-brain J. Pharmacol. Exp. Ther. 1989, 251, 888 893
  44. 44
    Klotz, K. N.; Lohse, M. J.; Schwabe, U.; Cristalli, G.; Vittori, S.; Grifantini, M. 2-Chloro-N-6-[H-3]cyclopentyladenosine ([H-3]Ccpa), a high-affinity agonist radioligand for A1 adenosine receptors Naunyn-Schmiedeberg's Arch. Pharmacol. 1989, 340, 679 683
  45. 45
    Olah, M. E.; Gallorodriguez, C.; Jacobson, K. A.; Stiles, G. L. I-125 4-aminobenzyl-5′-N-methylcarboxamidoadenosine, a high-affinity radioligand for the rat a(3) adenosine receptor Mol. Pharmacol. 1994, 45, 978 982
  46. 46
    Englert, M.; Quitterer, U.; Klotz, K. N. Effector coupling of stably transfected human A(3) adenosine receptors in CHO cells Biochem. Pharmacol. 2002, 64, 61 65
  47. 47
    Jacobson, K. A.; Park, K. S.; Jiang, J. L.; Kim, Y. C.; Olah, M. E.; Stiles, G. L.; Ji, X. D. Pharmacological characterization of novel A(3) adenosine receptor-selective antagonists Neuropharmacology 1997, 36, 1157 1165
  48. 48
    Nordstedt, C.; Fredholm, B. B. A modification of a protein-binding method for rapid quantification of camp in cell-culture supernatants and body-fluid Anal. Biochem. 1990, 189, 231 234
  49. 49
    Post, S. R.; Ostrom, R. S.; Insel, P. A. Biochemical methods for detection and measurement of cyclic AMP and adenylyl cyclase activity Methods Mol. Biol. 2000, 126, 363 374
  50. 50
    Bradford, M. M. Rapid and sensitive method for quantitation of microgram quantities of protein utilizing principle of protein−dye binding Anal. Biochem. 1976, 72, 248 254
  51. 51
    McGovern, S. L.; Helfand, B. T.; Feng, B.; Shoichet, B. K. A specific mechanism of nonspecific inhibition J. Med. Chem. 2003, 46, 4265 4272
  52. 52
    Kim, J. H.; Wess, J.; Vanrhee, A. M.; Schoneberg, T.; Jacobson, K. A. Site-directed mutagenesis identifies residues involved in ligand recognition in the human a(2a) adenosine receptor J. Biol. Chem. 1995, 270, 13987 13997
  53. 53
    Webb, T. R.; Lvovskiy, D.; Kim, S. A.; Ji, X. D.; Melman, N.; Linden, J.; Jacobson, K. A. Quinazolines as adenosine receptor antagonists: SAR and selectivity for A(2B) receptors Bioorg. Med. Chem. 2003, 11, 77 85
  54. 54
    Hert, J.; Irwin, J. J.; Laggner, C.; Keiser, M. J.; Shoichet, B. K. Quantifying biogenic bias in screening libraries Nat. Chem. Biol. 2009, 5, 479 483
  55. 55
    Soelaiman, S.; Wei, B. Q.; Bergson, P.; Lee, Y. S.; Shen, Y.; Mrksich, M.; Shoichet, B. K.; Tang, W. J. Structure-based inhibitor discovery against adenylyl cyclase toxins from pathogenic bacteria that cause anthrax and whooping cough J. Biol. Chem. 2003, 278, 25990 25997
  56. 56
    Katritch, V.; Jaakola, V. P.; Lane, J. R.; Lin, J.; Ijzerman, A. P.; Yeager, M.; Kufareva, I.; Stevens, R. C.; Abagyan, R. Structure-based discovery of novel chemotypes for adenosine A(2A) receptor antagonists J. Med. Chem. 2010, 53, 1799 1809

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 191 publications.

  1. Hyeongwoo Kim, Kyunghoon Lee, Chansu Kim, Jaechang Lim, Woo Youn Kim. DFRscore: Deep Learning-Based Scoring of Synthetic Complexity with Drug-Focused Retrosynthetic Analysis for High-Throughput Virtual Screening. Journal of Chemical Information and Modeling 2023, Article ASAP.
  2. Margarita Stampelou, Anna Suchankova, Efpraxia Tzortzini, Lakshiv Dhingra, Kerry Barkan, Nikolaos Lougiakis, Panagiotis Marakos, Nicole Pouli, Graham Ladds, Antonios Kolocouris. Dual A1/A3 Adenosine Receptor Antagonists: Binding Kinetics and Structure−Activity Relationship Studies Using Mutagenesis and Alchemical Binding Free Energy Calculations. Journal of Medicinal Chemistry 2022, 65 (19) , 13305-13327. https://doi.org/10.1021/acs.jmedchem.2c01123
  3. Vigneshwaran Namasivayam, Katja Silbermann, Michael Wiese, Jens Pahnke, Sven Marcel Stefan. C@PA: Computer-Aided Pattern Analysis to Predict Multitarget ABC Transporter Inhibitors. Journal of Medicinal Chemistry 2021, 64 (6) , 3350-3366. https://doi.org/10.1021/acs.jmedchem.0c02199
  4. Celeste N. Alverez, Jung-Eun Park, Kiran S. Toti, Yangliu Xia, Kristopher W. Krausz, Ganesha Rai, Jeong K. Bang, Frank J. Gonzalez, Kenneth A. Jacobson, Kyung S. Lee. Identification of a New Heterocyclic Scaffold for Inhibitors of the Polo-Box Domain of Polo-like Kinase 1. Journal of Medicinal Chemistry 2020, 63 (22) , 14087-14117. https://doi.org/10.1021/acs.jmedchem.0c01669
  5. Eleonora Comeo, Nicholas D. Kindon, Mark Soave, Leigh A. Stoddart, Laura E. Kilpatrick, Peter J. Scammells, Stephen J. Hill, Barrie Kellam. Subtype-Selective Fluorescent Ligands as Pharmacological Research Tools for the Human Adenosine A2A Receptor. Journal of Medicinal Chemistry 2020, 63 (5) , 2656-2672. https://doi.org/10.1021/acs.jmedchem.9b01856
  6. Flavio Ballante, Axel Rudling, Alexey Zeifman, Andreas Luttens, Duy Duc Vo, John J. Irwin, Jan Kihlberg, Jose Brea, Maria Isabel Loza, Jens Carlsson. Docking Finds GPCR Ligands in Dark Chemical Matter. Journal of Medicinal Chemistry 2020, 63 (2) , 613-620. https://doi.org/10.1021/acs.jmedchem.9b01560
  7. Panagiotis Lagarias, Kerry Barkan, Eva Tzortzini, Margarita Stampelou, Eleni Vrontaki, Graham Ladds, Antonios Kolocouris. Insights to the Binding of a Selective Adenosine A3 Receptor Antagonist Using Molecular Dynamic Simulations, MM-PBSA and MM-GBSA Free Energy Calculations, and Mutagenesis. Journal of Chemical Information and Modeling 2019, 59 (12) , 5183-5197. https://doi.org/10.1021/acs.jcim.9b00751
  8. Dahlia R. Weiss, Joel Karpiak, Xi-Ping Huang, Maria F. Sassano, Jiankun Lyu, Bryan L. Roth, Brian K. Shoichet. Selectivity Challenges in Docking Screens for GPCR Targets and Antitargets. Journal of Medicinal Chemistry 2018, 61 (15) , 6830-6845. https://doi.org/10.1021/acs.jmedchem.8b00718
  9. Mariama Jaiteh, Alexey Zeifman, Marcus Saarinen, Per Svenningsson, Jose Bréa, Maria Isabel Loza, Jens Carlsson. Docking Screens for Dual Inhibitors of Disparate Drug Targets for Parkinson’s Disease. Journal of Medicinal Chemistry 2018, 61 (12) , 5269-5278. https://doi.org/10.1021/acs.jmedchem.8b00204
  10. Panagiotis Lagarias, Eleni Vrontaki, George Lambrinidis, Dimitrios Stamatis, Marino Convertino, Gabriella Ortore, Thomas Mavromoustakos, Karl-Norbert Klotz, Antonios Kolocouris. Discovery of Novel Adenosine Receptor Antagonists through a Combined Structure- and Ligand-Based Approach Followed by Molecular Dynamics Investigation of Ligand Binding Mode. Journal of Chemical Information and Modeling 2018, 58 (4) , 794-815. https://doi.org/10.1021/acs.jcim.7b00455
  11. Yoonji Lee, Shaherin Basith, and Sun Choi . Recent Advances in Structure-Based Drug Design Targeting Class A G Protein-Coupled Receptors Utilizing Crystal Structures and Computational Simulations. Journal of Medicinal Chemistry 2018, 61 (1) , 1-46. https://doi.org/10.1021/acs.jmedchem.6b01453
  12. Inbar Fish, Anne Stößel, Katrin Eitel, Celine Valant, Sabine Albold, Harald Huebner, Dorothee Möller, Mary J. Clark, Roger K. Sunahara, Arthur Christopoulos, Brian K. Shoichet, and Peter Gmeiner . Structure-Based Design and Discovery of New M2 Receptor Agonists. Journal of Medicinal Chemistry 2017, 60 (22) , 9239-9250. https://doi.org/10.1021/acs.jmedchem.7b01113
  13. Sheng Tian, Xu Wang, Linlang Li, Xiaohu Zhang, Youyong Li, Feng Zhu, Tingjun Hou, and Xuechu Zhen . Discovery of Novel and Selective Adenosine A2A Receptor Antagonists for Treating Parkinson’s Disease through Comparative Structure-Based Virtual Screening. Journal of Chemical Information and Modeling 2017, 57 (6) , 1474-1487. https://doi.org/10.1021/acs.jcim.7b00188
  14. Marta Arimont, Shan-Liang Sun, Rob Leurs, Martine Smit, Iwan J. P. de Esch, and Chris de Graaf . Structural Analysis of Chemokine Receptor–Ligand Interactions. Journal of Medicinal Chemistry 2017, 60 (12) , 4735-4779. https://doi.org/10.1021/acs.jmedchem.6b01309
  15. Zhong Zheng, Xi-Ping Huang, Thomas J. Mangano, Rodger Zou, Xin Chen, Saheem A. Zaidi, Bryan L. Roth, Raymond C. Stevens, and Vsevolod Katritch . Structure-Based Discovery of New Antagonist and Biased Agonist Chemotypes for the Kappa Opioid Receptor. Journal of Medicinal Chemistry 2017, 60 (7) , 3070-3081. https://doi.org/10.1021/acs.jmedchem.7b00109
  16. Anirudh Ranganathan, Philipp Heine, Axel Rudling, Andreas Plückthun, Lutz Kummer, and Jens Carlsson . Ligand Discovery for a Peptide-Binding GPCR by Structure-Based Screening of Fragment- and Lead-Like Chemical Libraries. ACS Chemical Biology 2017, 12 (3) , 735-745. https://doi.org/10.1021/acschembio.6b00646
  17. Thomas M. Frimurer, Franziska Mende, Anne-Sofie Graae, Maja S. Engelstoft, Kristoffer L. Egerod, Rie Nygaard, Lars-Ole Gerlach, Jakob Bondo Hansen, Thue W. Schwartz, and Birgitte Holst . Model-Based Discovery of Synthetic Agonists for the Zn2+-Sensing G-Protein-Coupled Receptor 39 (GPR39) Reveals Novel Biological Functions. Journal of Medicinal Chemistry 2017, 60 (3) , 886-898. https://doi.org/10.1021/acs.jmedchem.6b00648
  18. Ali Jazayeri, Stephen P. Andrews, and Fiona H. Marshall . Structurally Enabled Discovery of Adenosine A2A Receptor Antagonists. Chemical Reviews 2017, 117 (1) , 21-37. https://doi.org/10.1021/acs.chemrev.6b00119
  19. David Rodríguez, Saibal Chakraborty, Eugene Warnick, Steven Crane, Zhan-Guo Gao, Robert O’Connor, Kenneth A. Jacobson, and Jens Carlsson . Structure-Based Screening of Uncharted Chemical Space for Atypical Adenosine Receptor Agonists. ACS Chemical Biology 2016, 11 (10) , 2763-2772. https://doi.org/10.1021/acschembio.6b00357
  20. Ainoleena Turku, Alexandre Borrel, Teppo O. Leino, Lasse Karhu, Jyrki P. Kukkonen, and Henri Xhaard . Pharmacophore Model To Discover OX1 and OX2 Orexin Receptor Ligands. Journal of Medicinal Chemistry 2016, 59 (18) , 8263-8275. https://doi.org/10.1021/acs.jmedchem.6b00333
  21. John J. Irwin and Brian K. Shoichet . Docking Screens for Novel Ligands Conferring New Biology. Journal of Medicinal Chemistry 2016, 59 (9) , 4103-4120. https://doi.org/10.1021/acs.jmedchem.5b02008
  22. Yu Zhou, Jing Ma, Xingyu Lin, Xi-Ping Huang, Kaichun Wu, and Niu Huang . Structure-Based Discovery of Novel and Selective 5-Hydroxytryptamine 2B Receptor Antagonists for the Treatment of Irritable Bowel Syndrome. Journal of Medicinal Chemistry 2016, 59 (2) , 707-720. https://doi.org/10.1021/acs.jmedchem.5b01631
  23. Anirudh Ranganathan, Leigh A. Stoddart, Stephen J. Hill, and Jens Carlsson . Fragment-Based Discovery of Subtype-Selective Adenosine Receptor Ligands from Homology Models. Journal of Medicinal Chemistry 2015, 58 (24) , 9578-9590. https://doi.org/10.1021/acs.jmedchem.5b01120
  24. Teague Sterling and John J. Irwin . ZINC 15 – Ligand Discovery for Everyone. Journal of Chemical Information and Modeling 2015, 55 (11) , 2324-2337. https://doi.org/10.1021/acs.jcim.5b00559
  25. Albert J. Kooistra, Rob Leurs, Iwan J. P. de Esch, and Chris de Graaf . Structure-Based Prediction of G-Protein-Coupled Receptor Ligand Function: A β-Adrenoceptor Case Study. Journal of Chemical Information and Modeling 2015, 55 (5) , 1045-1061. https://doi.org/10.1021/acs.jcim.5b00066
  26. David Rodríguez, Zhang-Guo Gao, Steven M. Moss, Kenneth A. Jacobson, and Jens Carlsson . Molecular Docking Screening Using Agonist-Bound GPCR Structures: Probing the A2A Adenosine Receptor. Journal of Chemical Information and Modeling 2015, 55 (3) , 550-563. https://doi.org/10.1021/ci500639g
  27. Andrew Anighoro, Dagmar Stumpfe, Kathrin Heikamp, Kristin Beebe, Leonard M. Neckers, Jürgen Bajorath, and Giulio Rastelli . Computational Polypharmacology Analysis of the Heat Shock Protein 90 Interactome. Journal of Chemical Information and Modeling 2015, 55 (3) , 676-686. https://doi.org/10.1021/ci5006959
  28. Denis Schmidt, Viachaslau Bernat, Regine Brox, Nuska Tschammer, and Peter Kolb . Identifying Modulators of CXC Receptors 3 and 4 with Tailored Selectivity Using Multi-Target Docking. ACS Chemical Biology 2015, 10 (3) , 715-724. https://doi.org/10.1021/cb500577j
  29. Dahlia A. Goldfeld, Robert Murphy, Byungchan Kim, Lingle Wang, Thijs Beuming, Robert Abel, and Richard A. Friesner . Docking and Free Energy Perturbation Studies of Ligand Binding in the Kappa Opioid Receptor. The Journal of Physical Chemistry B 2015, 119 (3) , 824-835. https://doi.org/10.1021/jp5053612
  30. Andrew Anighoro, Jürgen Bajorath, and Giulio Rastelli . Polypharmacology: Challenges and Opportunities in Drug Discovery. Journal of Medicinal Chemistry 2014, 57 (19) , 7874-7887. https://doi.org/10.1021/jm5006463
  31. Antonella Ciancetta, Alberto Cuzzolin, and Stefano Moro . Alternative Quality Assessment Strategy to Compare Performances of GPCR-Ligand Docking Protocols: The Human Adenosine A2A Receptor as a Case Study. Journal of Chemical Information and Modeling 2014, 54 (8) , 2243-2254. https://doi.org/10.1021/ci5002857
  32. David Rodríguez, Anirudh Ranganathan, and Jens Carlsson . Strategies for Improved Modeling of GPCR-Drug Complexes: Blind Predictions of Serotonin Receptors Bound to Ergotamine. Journal of Chemical Information and Modeling 2014, 54 (7) , 2004-2021. https://doi.org/10.1021/ci5002235
  33. Eelke B. Lenselink, Thijs Beuming, Woody Sherman, Herman W. T. van Vlijmen, and Adriaan P. IJzerman . Selecting an Optimal Number of Binding Site Waters To Improve Virtual Screening Enrichments Against the Adenosine A2A Receptor. Journal of Chemical Information and Modeling 2014, 54 (6) , 1737-1746. https://doi.org/10.1021/ci5000455
  34. Dan Chen, Anirudh Ranganathan, Adriaan P. IJzerman, Gregg Siegal, and Jens Carlsson . Complementarity between in Silico and Biophysical Screening Approaches in Fragment-Based Lead Discovery against the A2A Adenosine Receptor. Journal of Chemical Information and Modeling 2013, 53 (10) , 2701-2714. https://doi.org/10.1021/ci4003156
  35. Magdalena Bacilieri, Antonella Ciancetta, Silvia Paoletta, Stephanie Federico, Sandro Cosconati, Barbara Cacciari, Sabrina Taliani, Federico Da Settimo, Ettore Novellino, Karl Norbert Klotz, Giampiero Spalluto, and Stefano Moro . Revisiting a Receptor-Based Pharmacophore Hypothesis for Human A2A Adenosine Receptor Antagonists. Journal of Chemical Information and Modeling 2013, 53 (7) , 1620-1637. https://doi.org/10.1021/ci300615u
  36. Yasushi Yoshikawa, Shinya Oishi, Tatsuhiko Kubo, Noriko Tanahara, Nobutaka Fujii, and Toshio Furuya . Optimized Method of G-Protein-Coupled Receptor Homology Modeling: Its Application to the Discovery of Novel CXCR7 Ligands. Journal of Medicinal Chemistry 2013, 56 (11) , 4236-4251. https://doi.org/10.1021/jm400307y
  37. Anne Stößel, Miriam Schlenk, Sonja Hinz, Petra Küppers, Jag Heer, Michael Gütschow, and Christa E. Müller . Dual Targeting of Adenosine A2A Receptors and Monoamine Oxidase B by 4H-3,1-Benzothiazin-4-ones. Journal of Medicinal Chemistry 2013, 56 (11) , 4580-4596. https://doi.org/10.1021/jm400336x
  38. Kenneth A. Jacobson . Structure-Based Approaches to Ligands for G-Protein-Coupled Adenosine and P2Y Receptors, from Small Molecules to Nanoconjugates. Journal of Medicinal Chemistry 2013, 56 (10) , 3749-3767. https://doi.org/10.1021/jm400422s
  39. Dahlia R. Weiss, SeungKirl Ahn, Maria F. Sassano, Andrew Kleist, Xiao Zhu, Ryan Strachan, Bryan L. Roth, Robert J. Lefkowitz, and Brian K. Shoichet . Conformation Guides Molecular Efficacy in Docking Screens of Activated β-2 Adrenergic G Protein Coupled Receptor. ACS Chemical Biology 2013, 8 (5) , 1018-1026. https://doi.org/10.1021/cb400103f
  40. John A. Christopher, Jason Brown, Andrew S. Doré, James C. Errey, Markus Koglin, Fiona H. Marshall, David G. Myszka, Rebecca L. Rich, Christopher G. Tate, Benjamin Tehan, Tony Warne, and Miles Congreve . Biophysical Fragment Screening of the β1-Adrenergic Receptor: Identification of High Affinity Arylpiperazine Leads Using Structure-Based Drug Design. Journal of Medicinal Chemistry 2013, 56 (9) , 3446-3455. https://doi.org/10.1021/jm400140q
  41. Ana Negri, Marie-Laure Rives, Michael J. Caspers, Thomas E. Prisinzano, Jonathan A. Javitch, and Marta Filizola . Discovery of a Novel Selective Kappa-Opioid Receptor Agonist Using Crystal Structure-Based Virtual Screening. Journal of Chemical Information and Modeling 2013, 53 (3) , 521-526. https://doi.org/10.1021/ci400019t
  42. Thijs Beuming and Woody Sherman . Current Assessment of Docking into GPCR Crystal Structures and Homology Models: Successes, Challenges, and Guidelines. Journal of Chemical Information and Modeling 2012, 52 (12) , 3263-3277. https://doi.org/10.1021/ci300411b
  43. Jens Carlsson, Dilip K. Tosh, Khai Phan, Zhan-Guo Gao, and Kenneth A. Jacobson . Structure–Activity Relationships and Molecular Modeling of 1,2,4-Triazoles as Adenosine Receptor Antagonists. ACS Medicinal Chemistry Letters 2012, 3 (9) , 715-720. https://doi.org/10.1021/ml300097g
  44. Michael M. Mysinger, Michael Carchia, John. J. Irwin, and Brian K. Shoichet . Directory of Useful Decoys, Enhanced (DUD-E): Better Ligands and Decoys for Better Benchmarking. Journal of Medicinal Chemistry 2012, 55 (14) , 6582-6594. https://doi.org/10.1021/jm300687e
  45. Christopher J. Langmead, Stephen P. Andrews, Miles Congreve, James C. Errey, Edward Hurrell, Fiona H. Marshall, Jonathan S. Mason, Christine M. Richardson, Nathan Robertson, Andrei Zhukov, and Malcolm Weir . Identification of Novel Adenosine A2A Receptor Antagonists by Virtual Screening. Journal of Medicinal Chemistry 2012, 55 (5) , 1904-1909. https://doi.org/10.1021/jm201455y
  46. Francesca Fanelli and Pier G. De Benedetti . Update 1 of: Computational Modeling Approaches to Structure–Function Analysis of G Protein-Coupled Receptors. Chemical Reviews 2011, 111 (12) , PR438-PR535. https://doi.org/10.1021/cr100437t
  47. Chris de Graaf, Albert J. Kooistra, Henry F. Vischer, Vsevolod Katritch, Martien Kuijer, Mitsunori Shiroishi, So Iwata, Tatsuro Shimamura, Raymond C. Stevens, Iwan J. P. de Esch, and Rob Leurs . Crystal Structure-Based Virtual Screening for Fragment-like Ligands of the Human Histamine H1 Receptor. Journal of Medicinal Chemistry 2011, 54 (23) , 8195-8206. https://doi.org/10.1021/jm2011589
  48. Miles Congreve, Christopher J. Langmead, Jonathan S. Mason, and Fiona H. Marshall . Progress in Structure Based Drug Design for G Protein-Coupled Receptors. Journal of Medicinal Chemistry 2011, 54 (13) , 4283-4311. https://doi.org/10.1021/jm200371q
  49. David Rodríguez, Ángel Piñeiro, and Hugo Gutiérrez-de-Terán . Molecular Dynamics Simulations Reveal Insights into Key Structural Elements of Adenosine Receptors. Biochemistry 2011, 50 (19) , 4194-4208. https://doi.org/10.1021/bi200100t
  50. Anette G. Sams, Gitte K. Mikkelsen, Mogens Larsen, Morten Langgård, Mark E. Howells, Tenna J. Schrøder, Lise T. Brennum, Lars Torup, Erling B. Jørgensen, Christoffer Bundgaard, Mads Kreilgård, and Benny Bang-Andersen . Discovery of Phosphoric Acid Mono-{2-[(E/Z)-4-(3,3-dimethyl-butyrylamino)-3,5-difluoro-benzoylimino]-thiazol-3-ylmethyl} Ester (Lu AA47070): A Phosphonooxymethylene Prodrug of a Potent and Selective hA2A Receptor Antagonist. Journal of Medicinal Chemistry 2011, 54 (3) , 751-764. https://doi.org/10.1021/jm1008659
  51. Sharangdhar S. Phatak, Edgar A. Gatica, and Claudio N. Cavasotto. Ligand-Steered Modeling and Docking: A Benchmarking Study in Class A G-Protein-Coupled Receptors. Journal of Chemical Information and Modeling 2010, 50 (12) , 2119-2128. https://doi.org/10.1021/ci100285f
  52. Miru Tang, Chang Wen, Jie Lin, Hongming Chen, Ting Ran. Discovery of novel A2AR antagonists through deep learning-based virtual screening. Artificial Intelligence in the Life Sciences 2023, 3 , 100058. https://doi.org/10.1016/j.ailsci.2023.100058
  53. Pierre Matricon, Anh TN. Nguyen, Duc Duy Vo, Jo-Anne Baltos, Mariama Jaiteh, Andreas Luttens, Stefanie Kampen, Arthur Christopoulos, Jan Kihlberg, Lauren Therese May, Jens Carlsson. Structure-based virtual screening discovers potent and selective adenosine A1 receptor antagonists. European Journal of Medicinal Chemistry 2023, 257 , 115419. https://doi.org/10.1016/j.ejmech.2023.115419
  54. Haruki Yamane, Takashi Ishida. Helix encoder: a compound-protein interaction prediction model specifically designed for class A GPCRs. Frontiers in Bioinformatics 2023, 3 https://doi.org/10.3389/fbinf.2023.1193025
  55. Chia-Ju Hsieh, Sam Giannakoulias, E. James Petersson, Robert H. Mach. Computational Chemistry for the Identification of Lead Compounds for Radiotracer Development. Pharmaceuticals 2023, 16 (2) , 317. https://doi.org/10.3390/ph16020317
  56. Ashraf Ahmed Ali Abdusalam. In-silico identification of novel inhibitors for human Aurora kinase B form the ZINC database using molecular docking-based virtual screening. Research Results in Pharmacology 2022, 8 (4) , 89-99. https://doi.org/10.3897/rrpharmacology.8.82977
  57. Kenneth A. Jacobson, Zhan‐Guo Gao, Pierre Matricon, Matthew T. Eddy, Jens Carlsson. Adenosine A 2A receptor antagonists: from caffeine to selective non‐xanthines. British Journal of Pharmacology 2022, 179 (14) , 3496-3511. https://doi.org/10.1111/bph.15103
  58. Miru Tang, Chang Wen, Lin Jie, Hongming Chen, Ting Ran. Discovery of Novel A 2AR Antagonists Through Deep Learning-Based Virtual Screening. SSRN Electronic Journal 2022, 31 https://doi.org/10.2139/ssrn.4188435
  59. Wen-Ting Chu, Zhiqiang Yan, Xiakun Chu, Xiliang Zheng, Zuojia Liu, Li Xu, Kun Zhang, Jin Wang. Physics of biomolecular recognition and conformational dynamics. Reports on Progress in Physics 2021, 84 (12) , 126601. https://doi.org/10.1088/1361-6633/ac3800
  60. Flavio Ballante, Albert J Kooistra, Stefanie Kampen, Chris de Graaf, Jens Carlsson, . Structure-Based Virtual Screening for Ligands of G Protein–Coupled Receptors: What Can Molecular Docking Do for You?. Pharmacological Reviews 2021, 73 (4) , 1698-1736. https://doi.org/10.1124/pharmrev.120.000246
  61. Miles Congreve, John A. Christopher, Chris de Graaf. Structure‐Based Drug Design for G Protein‐Coupled Receptors. 2021, 1-59. https://doi.org/10.1002/0471266949.bmc269
  62. Mukuo Wang, Shujing Hou, Yu Wei, Dongmei Li, Jianping Lin, . Discovery of novel dual adenosine A1/A2A receptor antagonists using deep learning, pharmacophore modeling and molecular docking. PLOS Computational Biology 2021, 17 (3) , e1008821. https://doi.org/10.1371/journal.pcbi.1008821
  63. Bassam M. Ayoub, Haidy E. Michel, Shereen Mowaka, Moataz S. Hendy, Mariam M. Tadros. Repurposing of Omarigliptin as a Neuroprotective Agent Based on Docking with A2A Adenosine and AChE Receptors, Brain GLP-1 Response and Its Brain/Plasma Concentration Ratio after 28 Days Multiple Doses in Rats Using LC-MS/MS. Molecules 2021, 26 (4) , 889. https://doi.org/10.3390/molecules26040889
  64. Kalpana K. Bhanumathy, Omar Abuhussein, Frederick S. Vizeacoumar, Andrew Freywald, Franco J. Vizeacoumar, Christopher P. Phenix, Eric W. Price, Ran Cao. Computational Prediction of Chemical Tools for Identification and Validation of Synthetic Lethal Interaction Networks. 2021, 333-358. https://doi.org/10.1007/978-1-0716-1740-3_18
  65. Veronica Salmaso, Kenneth A. Jacobson. Purinergic Signaling: Impact of GPCR Structures on Rational Drug Design. ChemMedChem 2020, 15 (21) , 1958-1973. https://doi.org/10.1002/cmdc.202000465
  66. Miles Congreve, Chris de Graaf, Nigel A. Swain, Christopher G. Tate. Impact of GPCR Structures on Drug Discovery. Cell 2020, 181 (1) , 81-91. https://doi.org/10.1016/j.cell.2020.03.003
  67. Reed M. Stein, Hye Jin Kang, John D. McCorvy, Grant C. Glatfelter, Anthony J. Jones, Tao Che, Samuel Slocum, Xi-Ping Huang, Olena Savych, Yurii S. Moroz, Benjamin Stauch, Linda C. Johansson, Vadim Cherezov, Terry Kenakin, John J. Irwin, Brian K. Shoichet, Bryan L. Roth, Margarita L. Dubocovich. Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 2020, 579 (7800) , 609-614. https://doi.org/10.1038/s41586-020-2027-0
  68. Mariama Jaiteh, Ismael Rodríguez-Espigares, Jana Selent, Jens Carlsson, . Performance of virtual screening against GPCR homology models: Impact of template selection and treatment of binding site plasticity. PLOS Computational Biology 2020, 16 (3) , e1007680. https://doi.org/10.1371/journal.pcbi.1007680
  69. Cuihua Zhang, Qunlin Li, Lingwei Meng, Yujie Ren. Design of novel dopamine D 2 and serotonin 5-HT 2A receptors dual antagonists toward schizophrenia: An integrated study with QSAR, molecular docking, virtual screening and molecular dynamics simulations. Journal of Biomolecular Structure and Dynamics 2020, 38 (3) , 860-885. https://doi.org/10.1080/07391102.2019.1590244
  70. Yu Wei, Mukuo Wang, Yang Li, Zhangyong Hong, Dongmei Li, Jianping Lin. Identification of new potent A1 adenosine receptor antagonists using a multistage virtual screening approach. European Journal of Medicinal Chemistry 2020, 187 , 111936. https://doi.org/10.1016/j.ejmech.2019.111936
  71. Francesca Deflorian, Jonathan S. Mason, Andrea Bortolato, Benjamin G. Tehan. Impact of Recently Determined Crystallographic Structures of GPCRs on Drug Discovery. 2020, 449-477. https://doi.org/10.1002/9781118681121.ch19
  72. Xiangli Qu, Dejian Wang, Beili Wu. Progress in GPCR structure determination. 2020, 3-22. https://doi.org/10.1016/B978-0-12-816228-6.00001-5
  73. Jinan Wang, Apurba Bhattarai, Waseem Imtiaz Ahmad, Treyton S. Farnan, Karen Priyadarshini John, Yinglong Miao. Computer-aided GPCR drug discovery. 2020, 283-293. https://doi.org/10.1016/B978-0-12-816228-6.00015-5
  74. Sumit Jamwal, Ashish Mittal, Puneet Kumar, Dana M. Alhayani, Amal Al-Aboudi. Therapeutic Potential of Agonists and Antagonists of A1, A2a, A2b and A3 Adenosine Receptors. Current Pharmaceutical Design 2019, 25 (26) , 2892-2905. https://doi.org/10.2174/1381612825666190716112319
  75. Omar H.A. Al-Attraqchi, Mahesh Attimarad, Katharigatta N. Venugopala, Anroop Nair, Noor H.A. Al-Attraqchi. Adenosine A2A Receptor as a Potential Drug Target - Current Status and Future Perspectives. Current Pharmaceutical Design 2019, 25 (25) , 2716-2740. https://doi.org/10.2174/1381612825666190716113444
  76. Pabitra Narayan Samanta, Supratik Kar, Jerzy Leszczynski. Recent Advances of In-Silico Modeling of Potent Antagonists for the Adenosine Receptors. Current Pharmaceutical Design 2019, 25 (7) , 750-773. https://doi.org/10.2174/1381612825666190304123545
  77. Nikhil Agrawal, Balakumar Chandrasekaran, Amal Al-Aboudi. Recent Advances in the In-silico Structure-based and Ligand-based Approaches for the Design and Discovery of Agonists and Antagonists of A2A Adenosine Receptor. Current Pharmaceutical Design 2019, 25 (7) , 774-782. https://doi.org/10.2174/1381612825666190306162006
  78. H. T. Zhu, L. Y. Qin, T. Liu, Y. Luo. A Convenient Synthesis of N2-Alkylated Guanines. Russian Journal of Organic Chemistry 2019, 55 (6) , 874-878. https://doi.org/10.1134/S1070428019060198
  79. Rita C. Acúrcio, Anna Scomparin, Ronit Satchi‐Fainaro, Helena F. Florindo, Rita C. Guedes. Computer‐aided drug design in new druggable targets for the next generation of immune‐oncology therapies. WIREs Computational Molecular Science 2019, 9 (3) https://doi.org/10.1002/wcms.1397
  80. Guillaume Ferré, Georges Czaplicki, Pascal Demange, Alain Milon. Structure and dynamics of dynorphin peptide and its receptor. 2019, 17-47. https://doi.org/10.1016/bs.vh.2019.05.006
  81. Miles Congreve, Giles A. Brown, Alexandra Borodovsky, Michelle L. Lamb. Targeting adenosine A 2A receptor antagonism for treatment of cancer. Expert Opinion on Drug Discovery 2018, 13 (11) , 997-1003. https://doi.org/10.1080/17460441.2018.1534825
  82. Shaherin Basith, Minghua Cui, Stephani J. Y. Macalino, Jongmi Park, Nina A. B. Clavio, Soosung Kang, Sun Choi. Exploring G Protein-Coupled Receptors (GPCRs) Ligand Space via Cheminformatics Approaches: Impact on Rational Drug Design. Frontiers in Pharmacology 2018, 9 https://doi.org/10.3389/fphar.2018.00128
  83. Magdalena Korczynska, Mary J. Clark, Celine Valant, Jun Xu, Ee Von Moo, Sabine Albold, Dahlia R. Weiss, Hayarpi Torosyan, Weijiao Huang, Andrew C. Kruse, Brent R. Lyda, Lauren T. May, Jo-Anne Baltos, Patrick M. Sexton, Brian K. Kobilka, Arthur Christopoulos, Brian K. Shoichet, Roger K. Sunahara. Structure-based discovery of selective positive allosteric modulators of antagonists for the M 2 muscarinic acetylcholine receptor. Proceedings of the National Academy of Sciences 2018, 115 (10) https://doi.org/10.1073/pnas.1718037115
  84. Khushal Kapadiya, Yashwantsinh Jadeja, Ranjan Khunt. Synthesis of Purine‐based Triazoles by Copper (I)‐catalyzed Huisgen Azide–Alkyne Cycloaddition Reaction. Journal of Heterocyclic Chemistry 2018, 55 (1) , 199-208. https://doi.org/10.1002/jhet.3025
  85. Agostinho Lemos, Rita Melo, Irina S. Moreira, M. Natália D. S. Cordeiro. Computer-Aided Drug Design Approaches to Study Key Therapeutic Targets in Alzheimer’s Disease. 2018, 61-106. https://doi.org/10.1007/978-1-4939-7404-7_3
  86. Antonella Ciancetta, Kenneth A. Jacobson. Breakthrough in GPCR Crystallography and Its Impact on Computer-Aided Drug Design. 2018, 45-72. https://doi.org/10.1007/978-1-4939-7465-8_3
  87. Stefania Baraldi, Pier Giovanni Baraldi, Paola Oliva, Kiran S. Toti, Antonella Ciancetta, Kenneth A. Jacobson. A2A Adenosine Receptor: Structures, Modeling, and Medicinal Chemistry. 2018, 91-136. https://doi.org/10.1007/978-3-319-90808-3_5
  88. Eric Rouviere, Clément Arnarez, Lewen Yang, Edward Lyman. Identification of Two New Cholesterol Interaction Sites on the A2A Adenosine Receptor. Biophysical Journal 2017, 113 (11) , 2415-2424. https://doi.org/10.1016/j.bpj.2017.09.027
  89. Mette Trauelsen, Elisabeth Rexen Ulven, Siv A. Hjorth, Matjaz Brvar, Claudia Monaco, Thomas M. Frimurer, Thue W. Schwartz. Receptor structure-based discovery of non-metabolite agonists for the succinate receptor GPR91. Molecular Metabolism 2017, 6 (12) , 1585-1596. https://doi.org/10.1016/j.molmet.2017.09.005
  90. Pierre Matricon, Anirudh Ranganathan, Eugene Warnick, Zhan-Guo Gao, Axel Rudling, Catia Lambertucci, Gabriella Marucci, Aitakin Ezzati, Mariama Jaiteh, Diego Dal Ben, Kenneth A. Jacobson, Jens Carlsson. Fragment optimization for GPCRs by molecular dynamics free energy calculations: Probing druggable subpockets of the A 2A adenosine receptor binding site. Scientific Reports 2017, 7 (1) https://doi.org/10.1038/s41598-017-04905-0
  91. Phillip T. Lowe, Sergio Dall'Angelo, Thea Mulder‐Krieger, Adriaan P. IJzerman, Matteo Zanda, David O'Hagan. A New Class of Fluorinated A 2A Adenosine Receptor Agonist with Application to Last‐Step Enzymatic [ 18 F]Fluorination for PET Imaging. ChemBioChem 2017, 18 (21) , 2156-2164. https://doi.org/10.1002/cbic.201700382
  92. Bryan L Roth, John J Irwin, Brian K Shoichet. Discovery of new GPCR ligands to illuminate new biology. Nature Chemical Biology 2017, 13 (11) , 1143-1151. https://doi.org/10.1038/nchembio.2490
  93. Sheng Wang, Daniel Wacker, Anat Levit, Tao Che, Robin M. Betz, John D. McCorvy, A. J. Venkatakrishnan, Xi-Ping Huang, Ron O. Dror, Brian K. Shoichet, Bryan L. Roth. D 4 dopamine receptor high-resolution structures enable the discovery of selective agonists. Science 2017, 358 (6361) , 381-386. https://doi.org/10.1126/science.aan5468
  94. Valentina Abet, Fabiana Filace, Javier Recio, Julio Alvarez-Builla, Carolina Burgos. Prodrug approach: An overview of recent cases. European Journal of Medicinal Chemistry 2017, 127 , 810-827. https://doi.org/10.1016/j.ejmech.2016.10.061
  95. Anirudh Ranganathan, David Rodríguez, Jens Carlsson. Structure-Based Discovery of GPCR Ligands from Crystal Structures and Homology Models. 2017, 65-99. https://doi.org/10.1007/7355_2016_25
  96. M. Congreve, A. Bortolato, G. Brown, R.M. Cooke. Modeling and Design for Membrane Protein Targets. 2017, 145-188. https://doi.org/10.1016/B978-0-12-409547-2.12358-3
  97. Eelke B. Lenselink, Thijs Beuming, Corine van Veen, Arnault Massink, Woody Sherman, Herman W. T. van Vlijmen, Adriaan P. IJzerman. In search of novel ligands using a structure-based approach: a case study on the adenosine A2A receptor. Journal of Computer-Aided Molecular Design 2016, 30 (10) , 863-874. https://doi.org/10.1007/s10822-016-9963-7
  98. Aashish Manglik, Henry Lin, Dipendra K. Aryal, John D. McCorvy, Daniela Dengler, Gregory Corder, Anat Levit, Ralf C. Kling, Viachaslau Bernat, Harald Hübner, Xi-Ping Huang, Maria F. Sassano, Patrick M. Giguère, Stefan Löber, Da Duan, Grégory Scherrer, Brian K. Kobilka, Peter Gmeiner, Bryan L. Roth, Brian K. Shoichet. Structure-based discovery of opioid analgesics with reduced side effects. Nature 2016, 537 (7619) , 185-190. https://doi.org/10.1038/nature19112
  99. Celine Lacroix, Inbar Fish, Hayarpi Torosyan, Pranavan Parathaman, John J. Irwin, Brian K. Shoichet, Stephane Angers, . Identification of Novel Smoothened Ligands Using Structure-Based Docking. PLOS ONE 2016, 11 (8) , e0160365. https://doi.org/10.1371/journal.pone.0160365
  100. Elham Safarzadeh, Farhad Jadidi-Niaragh, Morteza Motallebnezhad, Mehdi Yousefi. The role of adenosine and adenosine receptors in the immunopathogenesis of multiple sclerosis. Inflammation Research 2016, 65 (7) , 511-520. https://doi.org/10.1007/s00011-016-0936-z
Load all citations
  • Abstract

    Chart 1

    Chart 1. Structures of Known Agonists (13) and Antagonists (46) of the A2A Adenosine Receptor

    Figure 1

    Figure 1. Binding mode of the cocrystallized ligand 6 (A) and the predicted binding modes of the seven ligands discovered in the docking screen (B−H). The A2A AR binding site is shown in white ribbons with the side chains of Glu169 and Asn253 in sticks. In (A) the cocrystallized ligand 6 is shown using orange carbon atoms. In (B−H), the crystallographic ligand is shown using blue lines and the docking poses for the ligands are depicted with orange carbon atoms. Black dotted lines indicate hydrogen bonds. The compounds are (B) 7, (C) 8, (D) 9, (E) 10, (F) 11, (G) 12, and (H) 13.

    Figure 2

    Figure 2. Representative dose−response curves for displacement of binding of the radiolabeled A2A AR agonist 3 by compounds 9, 10, and 11.

    Figure 3

    Figure 3. Functional assay based on measuring the production of cAMP for 3 (control), a potent A2A AR agonist, with or without 10 μM 9 or 11. The dose−response curve is shifted for both compounds, as expected in the case of competitive antagonistic inhibition. The % activation refers to production of cAMP normalized to the effect of 3 at 100 μM.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 56 other publications.

    1. 1
      Overington, J. P.; Al-Lazikani, B.; Hopkins, A. L. How many drug targets are there? Nat. Rev. Drug Discovery 2006, 5, 993 996
    2. 2
      Cherezov, V.; Rosenbaum, D. M.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Kuhn, P.; Weis, W. I.; Kobilka, B. K.; Stevens, R. C. High-resolution crystal structure of an engineered human beta(2)-adrenergic G protein-coupled receptor Science 2007, 318, 1258 1265
    3. 3
      Rosenbaum, D. M.; Cherezov, V.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Yao, X. J.; Weis, W. I.; Stevens, R. C.; Kobilka, B. K. GPCR engineering yields high-resolution structural insights into beta(2)-adrenergic receptor function Science 2007, 318, 1266 1273
    4. 4
      Warne, T.; Serrano-Vega, M. J.; Baker, J. G.; Moukhametzianov, R.; Edwards, P. C.; Henderson, R.; Leslie, A. G. W.; Tate, C. G.; Schertler, G. F. X. Structure of a beta(1)-adrenergic G-protein-coupled receptor Nature 2008, 454, 486 491
    5. 5
      Jaakola, V. P.; Griffith, M. T.; Hanson, M. A.; Cherezov, V.; Chien, E. Y. T.; Lane, J. R.; IJzerman, A. P.; Stevens, R. C. The 2.6 angstrom crystal structure of a human A(2A) adenosine receptor bound to an antagonist Science 2008, 322, 1211 1217
    6. 6
      Congreve, M.; Marshall, F. The impact of GPCR structures on pharmacology and structure-based drug design Br. J. Pharmacol. 2010, 159, 986 996
    7. 7
      Moro, S.; Gao, Z. G.; Jacobson, K. A.; Spalluto, G. Progress in the pursuit of therapeutic adenosine receptor antagonists Med. Res. Rev. 2006, 26, 131 159
    8. 8
      Jacobson, K. A.; Gao, Z. G. Adenosine receptors as therapeutic targets Nat. Rev. Drug Discovery 2006, 5, 247 264
    9. 9
      Sebastiao, A. M.; Ribeiro, J. A. Adenosine receptors and the central nervous system Handb. Exp. Pharmacol. 2009, 471 534
    10. 10
      Blackburn, M. R.; Vance, C. O.; Morschl, E.; Wilson, C. N. Adenosine receptors and inflammation Handb. Exp. Pharmacol. 2009, 215 269
    11. 11
      Cristalli, G.; Muller, C. E.; Volpini, R. Recent developments in adenosine A2A receptor ligands Handb. Exp. Pharmacol. 2009, 59 98
    12. 12
      Poucher, S. M.; Keddie, J. R.; Singh, P.; Stoggall, S. M.; Caulkett, P. W. R.; Jones, G.; Collis, M. G. The in-vitro pharmacology of Zm-241385, a potent, nonxanthine, a(2a) selective adenosine receptor antagonist Br. J. Pharmacol. 1995, 115, 1096 1102
    13. 13
      Degen, J.; Rarey, M. FlexNovo: structure-based searching in large fragment spaces ChemMedChem 2006, 1, 854 868
    14. 14
      Jones, G.; Willett, P.; Glen, R. C.; Leach, A. R.; Taylor, R. Development and validation of a genetic algorithm for flexible docking J. Mol. Biol. 1997, 267, 727 748
    15. 15
      Kairys, V.; Fernandes, M. X.; Gilson, M. K. Screening drug-like compounds by docking to homology models: a systematic study J. Chem. Inf. Model. 2006, 46, 365 379
    16. 16
      Lorber, D. M.; Shoichet, B. K. Flexible ligand docking using conformational ensembles Protein Sci. 1998, 7, 938 950
    17. 17
      Lorber, D. M.; Shoichet, B. K. Hierarchical docking of databases of multiple ligand conformations Curr. Top. Med. Chem. 2005, 5, 739 749
    18. 18
      Zavodszky, M. I.; Kuhn, L. A. Side-chain flexibility in protein−ligand binding: the minimal rotation hypothesis Protein Sci. 2005, 14, 1104 1114
    19. 19
      Kolb, P.; Rosenbaum, D. M.; Irwin, J. J.; Fung, J. J.; Kobilka, B. K.; Shoichet, B. K. Structure-based discovery of beta(2)-adrenergic receptor ligands Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 6843 6848
    20. 20
      Sabio, M.; Jones, K.; Topiol, S. Use of the X-ray structure of the beta(2)-adrenergic receptor for drug discovery. Part 2: Identification of active compounds Bioorg. Med. Chem. Lett. 2008, 18, 5391 5395
    21. 21
      de Graaf, C.; Rognan, D. Selective structure-based virtual screening for full and partial agonists of the beta 2 adrenergic receptor J. Med. Chem. 2008, 51, 4978 4985
    22. 22
      Katritch, V.; Reynolds, K. A.; Cherezov, V.; Hanson, M. A.; Roth, C. B.; Yeager, M.; Abagyan, R. Analysis of full and partial agonists binding to beta(2)-adrenergic receptor suggests a role of transmembrane helix V in agonist-specific conformational changes J. Mol. Recognit. 2009, 22, 307 318
    23. 23
      Reynolds, K. A.; Katritch, V.; Abagyan, R. Identifying conformational changes of the beta(2) adrenoceptor that enable accurate prediction of ligand/receptor interactions and screening for GPCR modulators J. Comput.-Aided Mol. Des. 2009, 23, 273 288
    24. 24
      Kuntz, I. D.; Blaney, J. M.; Oatley, S. J.; Langridge, R.; Ferrin, T. E. A geometric approach to macromolecule−ligand interactions J. Mol. Biol. 1982, 161, 269 288
    25. 25
      Shoichet, B. K.; Kuntz, I. D. Matching chemistry and shape in molecular docking Protein Eng. 1993, 6, 723 732
    26. 26
      Nicholls, A.; Honig, B. A rapid finite-difference algorithm, utilizing successive over-relaxation to solve the Poisson−Boltzmann equation J. Comput. Chem. 1991, 12, 435 445
    27. 27
      Weiner, S. J.; Kollman, P. A.; Case, D. A.; Singh, U. C.; Ghio, C.; Alagona, G.; Profeta, S.; Weiner, P. A new force-field for molecular mechanical simulation of nucleic-acids and proteins J. Am. Chem. Soc. 1984, 106, 765 784
    28. 28
      Babaoglu, K.; Simeonov, A.; Lrwin, J. J.; Nelson, M. E.; Feng, B.; Thomas, C. J.; Cancian, L.; Costi, M. P.; Maltby, D. A.; Jadhav, A.; Inglese, J.; Austin, C. P.; Shoichet, B. K. Comprehensive mechanistic analysis of hits from high-throughput and docking screens against beta-lactamase J. Med. Chem. 2008, 51, 2502 2511
    29. 29
      Powers, R. A.; Morandi, F.; Shoichet, B. K. Structure-based discovery of a novel, noncovalent inhibitor of AmpC beta-lactamase Structure 2002, 10, 1013 1023
    30. 30
      Meng, E. C.; Shoichet, B. K.; Kuntz, I. D. Automated docking with grid-based energy evaluation J. Comput. Chem. 1992, 13, 505 524
    31. 31
      Shoichet, B. K.; Leach, A. R.; Kuntz, I. D. Ligand solvation in molecular docking Proteins: Struct., Funct., Genet. 1999, 34, 4 16
    32. 32
      Wei, B. Q. Q.; Baase, W. A.; Weaver, L. H.; Matthews, B. W.; Shoichet, B. K. A model binding site for testing scoring functions in molecular docking J. Mol. Biol. 2002, 322, 339 355
    33. 33
      Irwin, J. J.; Shoichet, B. K. ZINC—a free database of commercially available compounds for virtual screening J. Chem. Inf. Model. 2005, 45, 177 182
    34. 34
      Bostrom, J.; Greenwood, J. R.; Gottfries, J. Assessing the performance of OMEGA with respect to retrieving bioactive conformations J. Mol. Graphics Modell. 2003, 21, 449 462
    35. 35
      Chambers, C. C.; Hawkins, G. D.; Cramer, C. J.; Truhlar, D. G. Model for aqueous solvation based on class IV atomic charges and first solvation shell effects J. Phys. Chem. 1996, 100, 16385 16398
    36. 36
      Li, J. B.; Zhu, T. H.; Cramer, C. J.; Truhlar, D. G. New class IV charge model for extracting accurate partial charges from wave functions J. Phys. Chem. A 1998, 102, 1820 1831
    37. 37
      Weiner, S. J.; Kollman, P. A.; Nguyen, D. T.; Case, D. A. An all atom force-field for simulations of proteins and nucleic-acids J. Comput. Chem. 1986, 7, 230 252
    38. 38
      http://accelrys.com/products/scitegic/.
    39. 39
      Olah, M.; Mracec, M.; Ostopovici, L.; Rad, R.; Bora, A.; Hadaruga, N.; Olah, I.; Banda, M.; Simon, Z.; Mracec, M.; Oprea, T. I. WOMBAT: World of Molecular Bioactivity. In Chemoinformatics in Drug Discovery; Oprea, T. I., Ed.; Wiley-VCH: Weinheim, Germany, 2005; pp 221239.
    40. 40
      http://www.ebi.ac.uk/chembl.
    41. 41
      Keiser, M. J.; Roth, B. L.; Armbruster, B. N.; Ernsberger, P.; Irwin, J. J.; Shoichet, B. K. Relating protein pharmacology by ligand chemistry Nat. Biotechnol. 2007, 25, 197 206
    42. 42
      Tondi, D.; Morandi, F.; Bonnet, R.; Costi, M. P.; Shoichet, B. K. Structure-based optimization of a non-beta-lactam lead results in inhibitors that do not up-regulate beta-lactamase expression in cell culture J. Am. Chem. Soc. 2005, 127, 4632 4639
    43. 43
      Jarvis, M. F.; Schulz, R.; Hutchison, A. J.; Do, U. H.; Sills, M. A.; Williams, M. [H-3] Cgs-21680, a selective A2 adenosine receptor agonist directly labels A2-receptors in rat-brain J. Pharmacol. Exp. Ther. 1989, 251, 888 893
    44. 44
      Klotz, K. N.; Lohse, M. J.; Schwabe, U.; Cristalli, G.; Vittori, S.; Grifantini, M. 2-Chloro-N-6-[H-3]cyclopentyladenosine ([H-3]Ccpa), a high-affinity agonist radioligand for A1 adenosine receptors Naunyn-Schmiedeberg's Arch. Pharmacol. 1989, 340, 679 683
    45. 45
      Olah, M. E.; Gallorodriguez, C.; Jacobson, K. A.; Stiles, G. L. I-125 4-aminobenzyl-5′-N-methylcarboxamidoadenosine, a high-affinity radioligand for the rat a(3) adenosine receptor Mol. Pharmacol. 1994, 45, 978 982
    46. 46
      Englert, M.; Quitterer, U.; Klotz, K. N. Effector coupling of stably transfected human A(3) adenosine receptors in CHO cells Biochem. Pharmacol. 2002, 64, 61 65
    47. 47
      Jacobson, K. A.; Park, K. S.; Jiang, J. L.; Kim, Y. C.; Olah, M. E.; Stiles, G. L.; Ji, X. D. Pharmacological characterization of novel A(3) adenosine receptor-selective antagonists Neuropharmacology 1997, 36, 1157 1165
    48. 48
      Nordstedt, C.; Fredholm, B. B. A modification of a protein-binding method for rapid quantification of camp in cell-culture supernatants and body-fluid Anal. Biochem. 1990, 189, 231 234
    49. 49
      Post, S. R.; Ostrom, R. S.; Insel, P. A. Biochemical methods for detection and measurement of cyclic AMP and adenylyl cyclase activity Methods Mol. Biol. 2000, 126, 363 374
    50. 50
      Bradford, M. M. Rapid and sensitive method for quantitation of microgram quantities of protein utilizing principle of protein−dye binding Anal. Biochem. 1976, 72, 248 254
    51. 51
      McGovern, S. L.; Helfand, B. T.; Feng, B.; Shoichet, B. K. A specific mechanism of nonspecific inhibition J. Med. Chem. 2003, 46, 4265 4272
    52. 52
      Kim, J. H.; Wess, J.; Vanrhee, A. M.; Schoneberg, T.; Jacobson, K. A. Site-directed mutagenesis identifies residues involved in ligand recognition in the human a(2a) adenosine receptor J. Biol. Chem. 1995, 270, 13987 13997
    53. 53
      Webb, T. R.; Lvovskiy, D.; Kim, S. A.; Ji, X. D.; Melman, N.; Linden, J.; Jacobson, K. A. Quinazolines as adenosine receptor antagonists: SAR and selectivity for A(2B) receptors Bioorg. Med. Chem. 2003, 11, 77 85
    54. 54
      Hert, J.; Irwin, J. J.; Laggner, C.; Keiser, M. J.; Shoichet, B. K. Quantifying biogenic bias in screening libraries Nat. Chem. Biol. 2009, 5, 479 483
    55. 55
      Soelaiman, S.; Wei, B. Q.; Bergson, P.; Lee, Y. S.; Shen, Y.; Mrksich, M.; Shoichet, B. K.; Tang, W. J. Structure-based inhibitor discovery against adenylyl cyclase toxins from pathogenic bacteria that cause anthrax and whooping cough J. Biol. Chem. 2003, 278, 25990 25997
    56. 56
      Katritch, V.; Jaakola, V. P.; Lane, J. R.; Lin, J.; Ijzerman, A. P.; Yeager, M.; Kufareva, I.; Stevens, R. C.; Abagyan, R. Structure-based discovery of novel chemotypes for adenosine A(2A) receptor antagonists J. Med. Chem. 2010, 53, 1799 1809
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    Table S1 of structures of the 500 top-ranking molecules from the docking screen. This material is available free of charge via the Internet at http://pubs.acs.org.


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect